throbber
Eur J Clin Pharmacol (2007) 63:677–686
`DOI 10.1007/s00228-007-0312-6
`
`PHARMACOKINETICS AND DISPOSITION
`
`The influence of hepatic impairment
`on the pharmacokinetics of the dipeptidyl peptidase IV
`(DPP-4) inhibitor vildagliptin
`
`Y.-L. He & R. Sabo & J. Campestrini & Y. Wang &
`M. Ligueros-Saylan & K. C. Lasseter & S. C. Dilzer &
`D. Howard & W. P. Dole
`
`Received: 8 January 2007 / Accepted: 4 April 2007 / Published online: 8 May 2007
`# Springer-Verlag 2007
`
`Abstract
`Objective Vildagliptin is a potent and selective dipeptidyl
`peptidase-IV (DPP-4) inhibitor that
`improves glycemic
`control
`in patients with type 2 diabetes mellitus by
`increasing α- and β-cell responsiveness to glucose. This
`study investigated the pharmacokinetics of vildagliptin in
`patients with hepatic impairment compared with healthy
`subjects.
`Methods This was an open-label, parallel-group study in
`patients with mild (n=6), moderate (n=6) or severe (n=4)
`hepatic impairment and healthy subjects (n=6). All subjects
`received a single 100-mg oral dose of vildagliptin, and
`plasma concentrations of vildagliptin and its main pharma-
`cologically inactive metabolite LAY151 were measured up
`to 36 h post-dose.
`Results Exposure to vildagliptin (AUC0–∞ and Cmax)
`decreased non-significantly by 20 and 30%, respectively,
`in patients with mild hepatic impairment [geometric mean
`
`Y.-L. He (*) : W. P. Dole
`Exploratory Development,
`Novartis Institutes for Biomedical Research, Inc.,
`400 Technology Square, Building 605, Rm 819,
`Cambridge, MA 02139-3584, USA
`e-mail: yanling.he@novartis.com
`R. Sabo : Y. Wang : M. Ligueros-Saylan : D. Howard
`Novartis Pharmaceuticals Corporation,
`East Hanover, NJ, USA
`
`J. Campestrini
`Novartis Pharma SA,
`Rueil-Malmaison, France
`K. C. Lasseter : S. C. Dilzer
`Pharmanet Development Group,
`Miami, FL, USA
`
`ratio (90% CI): AUC0–∞, 0.80 (0.60, 1.06), p=0.192; Cmax,
`0.70 (0.46, 1.05), p=0.149]. Exposure to vildagliptin was
`also decreased non-significantly in patients with moderate
`hepatic impairment [−8% for AUC0–∞, geometric mean ratio
`(90% CI): 0.92 (0.69, 1.23), p=0.630; −23% for Cmax,
`geometric mean ratio (90% CI): 0.77 (0.51, 1.17), p=0.293].
`In patients with severe hepatic impairment, Cmax was 6%
`lower than that in healthy subjects [geometric mean ratio
`(90% CI): 0.94 (0.59, 1.49), p=0.285], whereas AUC0–∞
`was increased by 22% [geometric mean ratio (90% CI): 1.22
`(0.89, 1.68), p=0.816). Across the hepatic impairment
`groups, LAY151 AUC0–∞ and Cmax were increased by 29–
`84% and 24–63%, respectively, compared with healthy
`subjects. The single 100-mg oral dose of vildagliptin was
`well tolerated by patients with hepatic impairment.
`Conclusions There was no significant difference in expo-
`sure to vildagliptin in patients with mild, moderate or
`severe hepatic impairment; therefore, no dose adjustment
`of vildagliptin is necessary in patients with hepatic
`impairment.
`
`Keywords Chronic liver disease . Dipeptidyl
`peptidase-IV inhibitor . Pharmacokinetics . Type 2 diabetes .
`Vildagliptin
`
`Introduction
`
`The incretin hormones, glucagon-like peptide-1 (GLP-1)
`and glucose-dependent insulinotropic polypeptide (GIP) are
`released from the gastrointestinal tract into the portal vein
`in response to food intake [1, 11]. GLP-1 and GIP are
`required for the maintenance of normal glucose tolerance,
`and they stimulate insulin release in a glucose-dependent
`
`Mylan EX 1013, Page 1
`
`

`

`678
`
`Eur J Clin Pharmacol (2007) 63:677–686
`
`manner [15, 21]. In healthy subjects, GLP-1 and GIP
`activity may be responsible for up to 70% of the insulin
`secreted in response to a meal [17]. In addition, GLP-1 also
`acts to suppress glucagon release [10]. Patients with type 2
`diabetes mellitus show impaired GLP-1 secretion, which
`may explain, at least in part, the reduced incretin effect seen
`in these patients [17, 22]. As GLP-1 is rapidly inactivated
`by the serine peptidase dipeptidyl peptidase IV (DPP-4) [5],
`a novel therapeutic approach to the treatment of type 2
`diabetes is to inhibit DPP-4 activity, and thereby prolong
`the physiological actions of GLP-1 [6].
`Vildagliptin (LAF237) is a potent, selective, orally active
`DPP-4 inhibitor developed for the treatment of type 2
`diabetes mellitus. In a clinical trial in patients with type 2
`diabetes, vildagliptin 100 mg once daily for 4 weeks
`increased post-prandial active GLP-1, reduced glucagon
`levels and improved glycemic control compared with
`placebo [3]. A recent study in drug-naive patients with type
`2 diabetes has also shown that vildagliptin improves
`pancreatic β-cell function by increasing the rate of insulin
`secretion [13]. Moreover, in a long-term study, the addition
`of vildagliptin 100 mg once daily to metformin therapy
`significantly improved glycemic control in patients with type
`2 diabetes mellitus over the 12 months of treatment [2].
`the
`Chronic liver disease can significantly impair
`function of hepatic drug-metabolizing enzymes (particular-
`ly microsomal oxidases), reduce hepatic blood flow and
`alter renal function [16, 19, 24]. The main metabolic
`pathway for vildagliptin in humans is hydrolysis to the
`inactive metabolite LAY151, which accounts for approxi-
`mately 69% of total elimination of an oral dose. Although
`85% of an oral dose of vildagliptin is ultimately excreted by
`the kidney, the liver is one of the major sites of vildagliptin
`metabolism (Novartis, data on file). Liver, kidney and
`intestinal microsomes are all capable of hydrolyzing
`vildagliptin to LAY151 in vitro. The aim of this study was
`to assess the effect of hepatic impairment on the disposition
`of vildagliptin by evaluating the pharmacokinetics, safety
`and tolerability of a single 100-mg oral dose of vildagliptin
`in patients with mild, moderate or severe hepatic impairment
`compared with healthy subjects with normal liver function.
`
`Methods
`
`Study design
`
`This was an open-label, single-dose, parallel-group study
`conducted at a single study center. Following a 21-day
`screening period, eligible subjects underwent baseline
`evaluation (Day −1) and received a single 100-mg oral dose
`of vildagliptin on Day 1. Blood samples were taken at
`regular intervals after dosing for the measurement of plasma
`
`drug levels. End-of-study evaluations were performed
`immediately after the last blood sample (36 h post-dose).
`Selection of the 100-mg dose of vildagliptin was based on
`the highest expected clinical dose. Treatment compliance
`was assured by the administration of vildagliptin under
`supervision of study center personnel and confirmed by the
`presence of vildagliptin in the plasma and urine.
`All subjects were admitted to the study center at least
`14 h prior to vildagliptin administration, and they were
`discharged following completion of the end-of-study evalua-
`tion. Vildagliptin was administered as a single tablet with 240
`mL of water between 7:00 and 8:30 A.M. following an
`overnight fast of at least 10 h. All subjects continued fasting
`for another 2 h after dosing, and unless performing a study
`assessment, subjects were required to rest quietly for a further
`4 h.
`The primary objective of this study was to assess the
`single-dose pharmacokinetics of vildagliptin 100 mg in
`patients with stable chronic liver disease compared with
`healthy subjects with normal liver function. A secondary
`objective was to assess the safety and tolerability of
`vildagliptin in patients with impaired hepatic function.
`
`Study population
`
`The study recruited men or women (ages: 18–60 years) of
`body weight ≥55 kg with a body mass index (BMI) of 22–
`40 kg/m2, resting pulse rate of 60–100 beats per minute and
`platelet count ≥50×109/L at screening and baseline. Female
`subjects had to be surgically sterile, postmenopausal, or use
`a double-barrier method of contraception.
`A total of 22 subjects were recruited, comprising healthy
`control subjects (n=6) and patients with mild (n=6),
`moderate (n=6) or severe (n=4) hepatic impairment. The
`diagnosis of cirrhosis was based on medical history and the
`presence of physical signs (liver firmness to palpitation,
`splenic enlargement, spider angioma, palmar erythema,
`parotid hypertrophy, testicular atrophy or gynecomastia).
`The Child-Pugh clinical assessment score was used to
`evaluate the degree of hepatic impairment based on
`guidance for clinical trials in patients with impaired hepatic
`function from the United States Food and Drug Adminis-
`tration [7]. Mild, moderate and severe hepatic impairment
`were defined by Child−Pugh assessment scores of 5–6, 7–9
`or 10–12, respectively (Table 1) [18].
`Apart from hepatic impairment, all participants were in
`good health, as determined by medical history, physical
`examination, vital signs, electrocardiogram (ECG) and
`laboratory tests at screening. Blood pressure (BP) inclusion
`criteria for healthy volunteers were a systolic BP (SBP) of
`90–160 mmHg and a diastolic BP (DBP) of 60–100 mmHg.
`For patients with hepatic impairment, inclusion criteria were
`an SBP of 90–180 mmHg and a DBP of 60–115 mmHg.
`
`Mylan EX 1013, Page 2
`
`

`

`Eur J Clin Pharmacol (2007) 63:677–686
`
`679
`
`Table 1 Child-Pugh classification scores for patients with mild,
`moderate or severe hepatic disease
`
`Mild hepatic
`impairment
`(n=6)
`
`Moderate
`hepatic
`impairment
`(n=6)
`
`Severe
`hepatic
`impairment
`(n=4)
`
`Overall
`Encephalopathy
`Ascites
`Bilirubin
`Albumin
`Prothrombin
`time
`
`6
`1–2
`1–2
`1
`1
`1
`
`7–8
`2
`2
`1–2
`1–2
`1
`
`10–12
`2
`1–3
`1–3
`2–3
`1–3
`
`Data are shown as minimum-maximum values for overall score and
`each component score
`
`Exclusion criteria for healthy volunteers included subjects
`who had used any prescription medication within 1 month
`prior to dosing or over-the-counter medications or vitamins
`within 14 days prior to dosing. Additional exclusion criteria
`for patients with hepatic impairment included: symptoms or
`history of Stage II (or worse) degree of encephalopathy within
`6 months of study entry; clinical evidence of severe ascites;
`history of surgical portosystemic shunt; prothrombin time >18
`s; any evidence of progressive liver disease within the
`previous 4 weeks, as
`indicated by changes in hepatic
`transaminases, alkaline phosphatase and γ-glutamyltransfer-
`ase, or a ≥50% worsening of serum bilirubin or prothrombin
`time. Patients with hepatic impairment were also excluded if,
`in the opinion of the study investigator,
`the degree of
`encephalopathy impaired the ability to provide written
`informed consent.
`Study participants were not permitted to engage in
`strenuous physical exercise for 7 days before dosing or to
`take alcohol for 72 h before dosing until after the study
`completion evaluation. Intake of xanthine-containing foods or
`beverages was discontinued 48 h before dosing and not
`permitted while participants were admitted to the study center.
`The study protocol was approved by the relevant local
`ethical
`review board. This study was conducted in
`accordance with the Guidelines for Good Clinical Practice
`and adhered to the ethical principles of the Declaration of
`Helsinki. All subjects provided written informed consent.
`
`Pharmacokinetic measurements
`
`Blood samples were taken by either direct venipuncture or
`by an indwelling cannula inserted in a forearm vein, at 0
`(pre-dose), 0.5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 10, 12, 15, 24 and
`36 h after vildagliptin administration. Blood samples were
`collected into sodium or lithium heparin tubes and stored at
`−70°C prior to analysis. Plasma samples were split equally
`for analysis of vildagliptin and LAY151. Urine samples
`
`were collected during the following time periods: pre-dose
`(overnight), 0–12, 12–24 and 24–36 h post-dose; urine
`collected during each time period was pooled, and a 5-mL
`aliquot was frozen.
`Plasma and urinary concentrations of vildagliptin and
`plasma concentrations of LAY151 were determined by liquid
`chromatography/mass spectrometry/mass spectrometry (LC/
`MS/MS) methods. All sample extractions were performed on
`10-mg Oasis HLB 96-well plates (Waters, Milford, Mass.)
`using an Automated Liquid Handling System model Multi
`probe II Plus HT EX. For the measurement of vildagliptin in
`plasma or urine, samples and internal standards were diluted
`in 4 mmol/L ammonium acetate (urine samples were also
`mixed with water). Samples were transferred to the
`extraction plates and successively washed with methanol:2%
`ammonium hydroxide (5:95, v/v), methanol:2% ammonium
`hydroxide (20:80, v/v) and water, and then eluted twice with
`methanol:0.1% trifluoroacetic acid (TFA; 80:20, v/v). Sam-
`ples were evaporated to a final volume of approximately
`50 μL and then diluted with methanol:0.5% ammonium
`hydroxide (15:85, v/v). For the measurement of LAY151 in
`plasma, samples and internal standards were mixed with
`4 mmol/L ammonium acetate. Samples were transferred to
`the extraction plates, successively washed with 0.1% TFA
`and 0.01% TFA and then eluted twice with methanol:0.5%
`ammonium acetate (80:20, v/v).
`The samples were analyzed by high-performance liquid
`chromatography (HPLC) using an XTerra MS C18 5 μm
`(150×2.1 mm) column (Waters) at 30°C with isocratic
`elution using 40% mobile phase A [10 mM ammonium
`acetate-methanol (95:5, v/v), pH 8] and 60% mobile phase B
`[acetonitrile-methanol (10/90, v/v)] at a flow rate of 0.2 mL/
`min. Detection was performed by MS/MS using an API3000
`(Applied Biosystems, Foster City, Calif.) or Quantum
`Discovery (Thermo Finnigan, San José, Calif.) mass
`spectrometer. The general conditions used were: positive
`ion mode with turbo ion spray or electro spray ionization.
`The masses for vildagliptin were precursor ion m/z 304 and
`product ion m/z 154; the masses for LAY151 were precursor
`m/z 323 and product m/z 173. For vildagliptin, the lower limit
`of quantification (LOQ) was 2 ng/mL in 0.2 mL of plasma and
`5 ng/mL in 0.1 mL of urine. The internal standard for
`vildagliptin was (2S)-1-[[(3-hydroxytricyclo[3.3.1.13,7]dec-
`1-yl)amino]acetyl]-2-pyrrolidine-2,3,4,5-13C-15N-carbonitrile-
`cyano-13C (Novartis Pharmaceuticals Corp, N.J.). Within-study
`assay validation showed an assay precision (co-efficient of
`variation) of 3.1–6.0% (bias of −4.2 to 3.5%) for plasma
`samples (nominal concentrations of 5.25, 400 and 900 ng/mL),
`and a precision of 6.5–7.4% (bias of −5.5 to 1.3%) for urine
`samples (nominal concentrations of 15, 2000 and 4000 ng/
`mL). For LAY151, the LOQ was 2 ng/mL in 0.2 mL of
`plasma. The internal standard was [D7] LAY151 (Novartis
`Pharma AG, Basel, Switzerland), and within-study assay
`
`Mylan EX 1013, Page 3
`
`

`

`680
`
`Eur J Clin Pharmacol (2007) 63:677–686
`
`validation showed a precision of 2.5–3.6% and a bias of −2.4
`to 5.9% (nominal concentrations of 5.01, 5.03, 406, 412, 902
`and 915 ng/mL). A single laboratory assayed all plasma and
`urine samples. Within-study assay validation was performed
`by an analysis of quality control samples.
`
`Safety and tolerability assessments
`
`Safety and tolerability assessments included the monitoring
`and recording of all adverse events (AEs), concomitant
`medications and significant non-drug therapies. Assessment
`of standard blood chemistries, hematological profile,
`urinalysis, physical examination, ECG and vital signs were
`performed at screening, baseline and at the end of the study.
`
`ranged from 0.25 to 0.29 for AUC and Cmax, respectively.
`Statistical comparisons of pharmacokinetic parameters be-
`tween the hepatic impairment groups and the healthy
`subjects group were performed for
`log-transformed
`AUC0–∞, AUC0–36 h and Cmax of vildagliptin using an
`analysis of variance (ANOVA) model. The source of
`variation included in the ANOVA model was hepatic group.
`Geometric mean ratios and corresponding 90% confidence
`intervals (CI) for each hepatic impairment group versus the
`healthy subjects group were calculated. Statistical compar-
`isons of pharmacokinetic parameters were performed using
`the PROC MIXED SAS procedure (SAS Institute, Cary, N.
`C.).
`
`Pharmacokinetic analyses
`
`Results
`
`The following pharmacokinetic parameters were calculated
`for vildagliptin using plasma concentration data: Cmax
`(maximum plasma concentration), tmax (time to reach Cmax)),
`AUC0–36 h (area under the plasma concentration–time curve
`for time period 0–36 h), AUC0–∞ (area under the plasma
`concentration-time curve extrapolated from time 0 to
`infinity),
`t1=2
`(elimination half-life associated with the
`terminal rate constant), CL/F (total clearance of drug from
`plasma, corrected for bioavailability F), Vz/F (apparent
`volume of distribution, corrected for bioavailability F) and
`CLR (renal clearance of drug from plasma). The total amount
`of vildagliptin excreted into urine was also measured. CLR
`was calculated using both plasma and urine data over the
`36-h assessment period. The following pharmacokinetic
`parameters were calculated for LAY151 using plasma
`concentration data: Cmax, tmax, AUC0–t, AUC0–∞ and t1=2
`.
`Pharmacokinetic parameters were obtained according to
`the non-compartmental methods using WINNONLIN PRO
`(ver. 4.0; Pharsight Corp., Mountain View, Calif.).
`
`Statistical analyses
`
`The sample size was determined mainly on the basis of the
`sensitivity of statistical testing to detect a clinically meaning-
`ful change in drug exposure between patients with hepatic
`impairment and healthy volunteers, and with reference to the
`sample sizes commonly used in pharmacokinetic studies in
`patients with hepatic impairment. Assuming an inter-subject
`co-efficient of variation (CV) of 0.25, a sample size of six in
`each of the three hepatic impairment patient subgroups and in
`the healthy subject group would ensure an 80% power to
`detect a 45% difference in pharmacokinetic parameter values
`between the healthy and hepatic-impaired groups, based on a
`two-sample, two-sided t-test at a 95% significance level.
`Previous vildagliptin pharmacokinetic studies in patients
`with type 2 diabetes mellitus showed that inter-subject CV
`
`Patient characteristics
`
`A total of 22 subjects were enrolled and completed the study
`(i.e. received vildagliptin and completed all post-dosing
`evaluations). Although the study originally planned to
`recruit six subjects in each group; only four subjects were
`enrolled in the severe hepatic impairment group due to the
`difficulty of recruiting these patients. Several patients were
`recruited with minor protocol deviations; one subject had a
`BMI (18.5 kg/m2) below the lower limit defined in the
`inclusion criteria; six participants were older than the
`maximum specified age of 60 years; one subject had a
`platelet count that was below that stated in the inclusion
`criteria. However, none of the deviations were considered
`significant by the investigator, and these subjects were
`allowed to participate in this study.
`Patients with hepatic impairment and healthy subjects
`showed similar baseline and demographic characteristics
`(Table 2), except that mean age was lower in the healthy
`subjects group (37.8 years) than in the hepatic impairment
`subgroups (54.8–56.5 years). The majority of patients with
`hepatic impairment had a medical history of alcoholism,
`which was not an active condition at the time of the study,
`or had existing medical conditions related to hepatic
`impairment (Table 2). Prothrombin time was increased in
`patients with mild, moderate or severe hepatic impairment
`compared with healthy subjects and was the longest in the
`severe hepatic impairment group. Total bilirubin was also
`markedly higher in patients with severe hepatic impairment
`than in patients in the other groups. Overall, ten (62.5%)
`patients with hepatic impairment were receiving medication
`for the treatment of hepatic disease or associated illnesses;
`the most common co-medication was spironolactone. There
`were no significant past or present medical conditions in the
`healthy volunteers group, and none of the healthy subjects
`were taking medications prior to enrolment.
`
`Mylan EX 1013, Page 4
`
`

`

`Eur J Clin Pharmacol (2007) 63:677–686
`
`Table 2 Baseline and demographic characteristics
`
`681
`
`Characteristic
`
`Healthy subjects (n=6)
`
`Patients with hepatic impairment
`
`Mild (n=6)
`
`Moderate (n=6)
`
`Severe (n=4)
`
`Age (years)
`Range
`Weight (kg)
`Height (cm)
`Gender, n (%)
`Male
`Female
`Ethnicity, n (%)
`Caucasian
`Black
`Othera
`Etiology of hepatic disease, n (%)
`Alcohol-induced liver cirrhosis
`Hepatitis C
`Clinical manifestations of hepatic disease
`Encephalopathy, n (%)
`Ascites, n (%)
`Peripheral edema, n (%)
`Albumin (g/dL)
`Total bilirubin (mg/dL)
`Prothrombin timeb (s)
`Creatinine clearancec (mL/min/1.73m2)
`
`37.8±10.9
`19–53
`70.3±9.4
`165±7
`
`3 (50)
`3 (50)
`
`0
`0
`6 (100)
`
`0
`0
`
`0
`0
`0
`4.4±0.1
`0.9±0.3
`11.8±0.4
`81.6±7.0
`
`56.0±5.8
`49–64
`79.2±16.2
`168±9
`
`3 (50)
`3 (50)
`
`4 (66.7)
`1 (16.7)
`1 (16.7)
`
`6 (100)
`3 (50)
`
`5 (83.3)
`4 (66.7)
`2 (33.3)
`4.3±0.2
`0.7±0.3
`12.1±0.8
`78.2±13.4
`
`56.5±6.6
`48–63
`80.5±17.8
`171±13
`
`5 (83)
`1 (17)
`
`1 (16.7)
`4 (66.7)
`1 (16.7)
`
`6 (100)
`3 (50)
`
`6 (100)
`4 (66.7)
`1 (16.7)
`3.8±0.5
`1.0±0.5
`12.2±1.1
`69.1±7.9
`
`54.8±14.7
`44–76
`72.8±20.3
`163±14
`
`2 (50)
`2 (50)
`
`1 (25)
`1 (25)
`2 (50)
`
`3 (75)
`4 (100)
`
`4 (100)
`4 (100)
`1 (25)
`3.0±0.6
`2.4±0.9
`16.1±2.6
`91.1±33.5
`
`Data are presented as mean±SD unless otherwise stated
`a ‘Other’ ethnic category includes ethnically Hispanic subjects of either Caucasian or black heritage
`b Prothrombin time was measured at screening
`c Calculated using the formula [(140–age in years) × weight in kg]/(serum creatinine × 72)
`
`Pharmacokinetics of vildagliptin in patients with mild,
`moderate or severe hepatic impairment
`
`The plasma concentration-time profiles for vildagliptin are
`illustrated in Fig. 1. The pharmacokinetic parameters for both
`vildagliptin and LAY151 are summarized in Table 3.
`Exposure to vildagliptin (AUC and Cmax) in patients with
`mild or moderate hepatic impairment was lower than in
`healthy subjects following the administration of a single 100-
`mg oral dose. In patients with mild hepatic impairment,
`vildagliptin AUC0–∞ and Cmax were reduced by 20% and 30,
`respectively, compared with healthy subjects [geometric mean
`ratio (90% CI): AUC0–∞, 0.80 (0.60, 1.06); Cmax, 0.70 (0.46,
`1.05)]. The reductions in AUC0–∞ and Cmax were not
`statistically significant compared with healthy subjects
`(p=
`0.192 and p=0.149, respectively). Vildagliptin AUC0–∞ was
`reduced by 8% and Cmax by 23% in patients with moderate
`hepatic impairment compared with healthy subjects [mean
`ratio (90% CI): AUC0–∞, 0.92 (0.69, 1.23); Cmax, 0.77 (0.51,
`1.17)]; however,
`these differences were not statistically
`significant (AUC0–∞, p=0.630 and Cmax, p=0.293 vs. healthy
`subjects). Plasma concentrations of vildagliptin following the
`administration of a single 100-mg oral dose were higher in
`patients with severe hepatic impairment than in healthy subjects
`
`Fig. 1 Plasma concentration−time profiles of vildagliptin in healthy
`subjects and patients with mild, moderate or severe hepatic impair-
`ment. Plasma concentrations of vildagliptin are shown after the
`administration of a single 100-mg oral dose to healthy subjects (n=6)
`and patients with mild (n=6), moderate (n=6) or severe (n=4) hepatic
`impairment. Data are presented as mean±SEM
`
`Mylan EX 1013, Page 5
`
`

`

`682
`
`Eur J Clin Pharmacol (2007) 63:677–686
`
`Table 3 Pharmacokinetic parameters of vildagliptin and LAY 151 in healthy subjects and patients with mild, moderate or severe hepatic
`impairment after administration of a single 100-mg oral dose
`
`Parameter
`
`Healthy subjects (n=6)
`
`Patients with hepatic impairment
`
`Vildagliptin
`tmax (h)
`Cmax (ng/mL)
`AUC0–t (ng·h/mL)
`AUC0–∞ ( ng·h/mL)
`t1=
`(h)
`2
`CL/F (L/h)
`Vz/F (L)
`CLR (L/h)
`LAY151
`tmax (h)
`Cmax (ng/mL)
`AUC0–t, (ng·h/mL)
`AUC0–∞ (ng·h/mL)
`t1=
`, h
`
`2
`
`1.3 (1.00–3.00)
`675±263
`2567±428
`2580±425
`2.0±0.5
`39.6±6.3
`115±32
`9.4±4.4
`
`6.0 (6.0–8.0)
`330±99
`5297±1715
`5641±1884
`7.9±0.7
`
`Mild (n=6)
`
`Moderate (n=6)
`
`Severe (n=4)
`
`Severe (n=3)a
`
`1.2 (1.0–2.0)
`497±229
`2076±514
`2101±512
`4.9±4.9
`50.4±14.1
`369±390
`10.2±2.2
`
`8.0 (6.0–10.0)
`406±108
`6721±1675
`7252±1782
`8.4±1.6
`
`1.0 (0.5–3.0)
`512±166
`2411±740
`2437±742
`3.1±1.6
`43.8±11.0
`200±122
`9.0±3.3
`
`9.00 (6.0–12.0)
`483±165
`7902±3168
`8396±3556
`7.2±0.8
`
`2.0 (1.0–4.0)
`632±247
`3322±1472
`3354±1462
`2.4±0.3
`34.4±15.0
`120±55
`6.2±4.0
`
`10.0 (8.0–12.0)
`549±217
`10160±4801
`11545±5964
`9.3±1.5
`
`1.5 (1.0–4.0)
`519±121
`2655±764
`2695±777
`2.4±0.3
`39.6±13.2
`138±50
`8.1±0.9
`
`10.0 (8.0–12.0)
`454±130
`8071±2897
`8893±3338
`8.7±1.0
`
`Data are presented as mean±SD, except for tmax, which is shown as median (minimum–maximum)
`a Omitting one subject who exhibited both hepatic and renal impairment (creatinine clearance: 43.8 mL/min/1.73 m2 )
`
`(Fig. 1, Table 3). Although Cmax and AUC0–∞ were 6% lower
`[geometric mean ratio (90% CI): 0.94 (0.59, 1.49)] and 22%
`higher [1.22 (0.89, 1.68)], respectively, in patients with severe
`hepatic impairment than in healthy subjects, these differences
`were not statistically significant (AUC0–∞, p=0.816; Cmax,
`p=0.285). Exposure to vildagliptin (AUC0–∞ and Cmax) in
`patients with severe hepatic impairment showed a greater
`degree of variability compared with the other groups (Fig. 2).
`Overall, variability in exposure to vildagliptin in patients with
`hepatic impairment appeared to increase with the severity of
`the hepatic impairment (Fig. 2). Changes in exposure were not
`consistent with the severity of hepatic impairment.
`It should be noted that one patient with severe hepatic
`impairment exhibited a vildagliptin AUC0–∞ of 5330 h ng/mL,
`which was considerably higher than the corresponding values
`for the other three patients in this subgroup (1829–3330 h ng/
`mL). Vildagliptin Cmax was also markedly higher in this
`patient
`than in the other patients with severe hepatic
`impairment (971 vs. 384–618 ng/mL, respectively). Moreover,
`CL/F and renal clearance (CLR) in this patient were 18.8 and
`0.33 L/h, respectively compared with values of 30.0–54.7
`(CL/F) and 7.2–9.0 L/h (CLR) for the other patients with
`severe hepatic impairment. This subject also exhibited a
`creatinine clearance of 44.4 mL/min/1.73 m2 compared with
`105.9–145.9 mL/min/1.73 m2 for other patients in the group.
`These observations suggest that this subject may have had an
`impairment of both the liver and the kidneys. Thus, the overall
`pharmacokinetic parameters in the severe hepatic impairment
`subgroup may have been skewed by the data from this patient.
`When these data were omitted from the analysis, exposure to
`vildagliptin in patients with severe hepatic impairment was
`
`actually reduced compared with healthy subjects and consis-
`tent with the trend observed in patients with mild or moderate
`hepatic impairment (Table 3).
`The mean CL/F of vildagliptin was 27% higher in
`patients with mild hepatic impairment
`than in healthy
`subjects, but it was similar in patients with moderate or
`severe hepatic impairment as compared with healthy
`subjects (Table 3). Mean CLR in patients with severe hepatic
`impairment was 34.4% lower than that in healthy subjects;
`however, this difference was minimal when one patient with
`moderate renal impairment (based on calculated creatinine
`clearance) was excluded (Table 3). Median tmax was
`comparable in patients with mild or moderate or severe
`hepatic impairment and healthy subjects, although the range
`for tmax was considerably larger in patients with severe
`hepatic impairment. Mean t1=2
`(4.9 h) appeared to be longer
`in patients with mild hepatic impairment than in healthy
`subjects (2.0 h) and in those with moderate or severe hepatic
`impairment (3.1 h). However, the mild hepatic impairment
`group had a large variability in estimated t1=2
`. with a CV of
`99% due to one subject who had measurable vildagliptin
`plasma concentrations at 36 h post-dose, whereas for all
`other patients, vildagliptin was not detectable in the plasma
`after 24 h.
`
`Pharmacokinetics of LAY151 in patients with mild,
`moderate or severe hepatic impairment
`
`The plasma concentration versus time profiles of LAY151 are
`illustrated in Fig. 3. Mean plasma concentrations of LAY151
`were higher in patients with mild or moderate hepatic
`
`Mylan EX 1013, Page 6
`
`

`

`Eur J Clin Pharmacol (2007) 63:677–686
`
`683
`
`Fig. 3 Plasma concentration–time profiles of LAY151 in healthy
`subjects and patients with mild, moderate or severe hepatic impair-
`ment. Plasma concentrations of LAY151 are shown after
`the
`administration of a single 100-mg oral dose of vildagliptin to healthy
`subjects (n=6) and patients with mild (n=6), moderate (n=6) or
`severe (n=4) hepatic impairment. Data are presented as mean±SEM
`
`impairment (8 and 9 h, respectively) than in healthy subjects
`(6 h; Table 3).
`Compared with healthy subjects, exposure to LAY151 was
`increased to a greater extent in patients with severe hepatic
`impairment than in patients with mild or moderate hepatic
`impairment (Table 3). AUC0–∞ and Cmax were increased by
`63% [geometric mean ratio (90% CI): 1.63 (1.14, 2.08)] and
`94% [mean ratio (90% CI): 1.94 (1.29, 2.91)], respectively,
`compared to healthy subjects. Median tmax was also longer in
`patients with severe hepatic impairment
`than in healthy
`subjects; however, mean t1=
`of LAY151 was comparable
`between the two groups (Table 3). It was notable that
`exposure to LAY151 was considerably higher in one patient
`(AUC0–∞, 19502 h ng/mL; Cmax, 834 ng/mL) than in the
`other
`three patients with severe hepatic impairment
`(AUC0–∞, 7,662–12,672 h ng/mL; Cmax, 411–600 ng/mL).
`This was the same subject who showed moderately impaired
`renal function and exhibited a markedly higher vildagliptin
`exposure than the remainder of the group. The pharmacoki-
`netic parameters of LAY151 in the severe hepatic impair-
`ment subgroup when data from this patient are omitted from
`the analysis are shown in Table 3.
`
`2
`
`Correlation between creatinine clearance and exposure
`to vildagliptin and LAY151
`
`The relationship between creatinine clearance and exposure
`to vildagliptin and LAY151 was assessed because the
`majority of an oral dose of vildagliptin is excreted in the
`
`Fig. 2 Lack of correlation between severity of hepatic impairment
`and (a) Cmax and (b) AUC0–∞ following the administration of a single
`100-mg oral dose of vildagliptin. Figure shows individual patients'
`Child-Pugh score and respective mean for (a) Cmax and (b) AUC0–∞
`for healthy subjects (n=6; filled circles) and patients with mild (n=6;
`open triangles), moderate (n=6; open squares) or severe (n=4; open
`diamonds) hepatic impairment
`
`than in healthy subjects (Table 3). LAY151
`impairment
`AUC0–∞ was 31% and 45% higher in patients with mild or
`moderate hepatic impairment, respectively, compared with
`healthy subjects [geometric mean ratio (90% CI): mild, 1.31
`(0.91, 1.89); moderate, 1.45 (1.01, 2.08)]. Furthermore,
`LAY151 Cmax was increased by 24% [mild hepatic
`impairment, mean ratio (90% CI): 1.24 (0.90, 1.71)] and
`45% [moderate hepatic impairment, mean ratio (90% CI):
`1.45 (1.05, 2.00)] compared with healthy subjects. The
`magnitude of the changes in LAY151 exposure appeared to
`be related to the severity of hepatic impairment. Median tmax
`was also longer in patients with mild or moderate hepatic
`
`Mylan EX 1013, Page 7
`
`

`

`684
`
`Eur J Clin Pharmacol (2007) 63:677–686
`
`urine. There was no significant correlation between creatinine
`clearance and vildagliptin AUC0–∞ (r=−0.246, p =0.270) or
`Cmax (−0.099, p=0.660; Fig. 4a). However, reductions in
`creatinine clearance showed a significant correlation with
`increased LAY151 AUC0–∞ (r=−0.475, p=0.026) and Cmax
`(r=−0.524, p= 0.012; Fig. 4b).
`
`Safety and tolerability
`
`Administration of a single 100-mg oral dose of vildagliptin
`was well tolerated both by patients with hepatic impairment
`
`Fig. 4 Correlation between creatinine clearance (CLcr) and AUC0–∞
`of (a) vildagliptin and (b) LAY151 following administration of a
`single 100-mg oral dose of vildagliptin. Figure shows individual
`patient creatinine clearance values and respective mean AUC0–∞
`values for (a) vildagliptin and (b) LAY151 for healthy subjects (n=6;
`filled circles) and patients with mild (n=6; open triangles), moderate
`(n=6; open squares) or severe (n=4; open diamonds) hepatic
`impairment
`
`and by healthy subjects. Overall, four (18.2%) of the 22
`participants reported a total of five AEs. One AE was
`reported each in the healthy subjects, and mild and
`moderate hepatic impairment groups;
`two AEs were
`reported in the severe hepatic impairment group. The most
`commonly reported AE was somnolence, which was
`reported by one patient each in the mild, moderate and
`severe hepatic impairment subgroups. There was also one
`report of diarrhea each in the healthy subject group and
`severe hepatic impairment group. All reported AEs were
`mild in intensity,
`transient and resolved spontaneously.
`There were no serious AEs.
`No clinically significant abnormalities other than those
`expected in patients with hepatic impairment were detected
`during the assessment of vital signs, blood chemistry,
`hematology or urinalysis.
`Vildagliptin had no notable effects on serum glucose
`levels in healthy volunteers or patients with hepatic
`impairment. In healthy volunteers, the mean serum glucose
`concentration at baselin

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket